Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 5161, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664315

ABSTRACT

Sonoporation via microbubble-mediated ultrasound exposure has shown potential in drug and gene delivery. However, there is a general lack of mechanistic knowledge on sonoporation-induced cellular impact after membrane resealing, and this issue has made it challenging to apply sonoporation efficiently in practice. Here, we present new evidence on how sonoporation, without endangering immediate cell viability, may disrupt downstream cellular hemostasis in ways that are distinguished from the bioeffects observed in other sonicated and unsonoporated cells. Sonoporation was realized on HL-60 leukemia cells by delivering pulsed ultrasound (1 MHz frequency, 0.50 MPa peak negative pressure; 10% duty cycle; 30 s exposure period; 29.1 J/cm2 acoustic energy density) in the presence of lipid-shelled microbubbles (1:1 cell-to-bubble ratio). Results showed that 54.6% of sonoporated cells, despite remaining initially viable, underwent apoptosis or necrosis at 24 h after sonoporation. Anti-proliferation behavior was also observed in sonoporated cells as their subpopulation size was reduced by 43.8% over 24 h. Preceding these cytotoxic events, the percentages of sonoporated cells in different cell cycle phases were found to be altered by 12 h after exposure. As well, for sonoporated cells, their expressions of cytoprotective genes in the heat shock protein-70 (HSP-70) family were upregulated by at least 4.1 fold at 3 h after exposure. Taken altogether, these findings indicate that sonoporated cells attempted to restore homeostasis after membrane resealing, but many of them ultimately failed to recover. Such mechanistic knowledge should be taken into account to devise more efficient sonoporation-mediated therapeutic protocols.


Subject(s)
Drug Delivery Systems , Gene Transfer Techniques , HSP72 Heat-Shock Proteins/genetics , Ultrasonic Waves , Cell Survival/radiation effects , Gene Expression/radiation effects , HL-60 Cells , HSP72 Heat-Shock Proteins/chemistry , HSP72 Heat-Shock Proteins/pharmacology , Humans , Lipids/chemistry , Lipids/pharmacology , Microbubbles/therapeutic use
2.
Cell Stress Chaperones ; 24(2): 461-467, 2019 03.
Article in English | MEDLINE | ID: mdl-30756293

ABSTRACT

The acute inflammatory response to active or passive activities that increase body temperature may aid to reduce chronic low-grade inflammation. This study investigates the impact of temperature and extracellular heat shock protein 72 (eHsp72) on the acute intracellular Hsp72 (iHsp72) and interleukin-6 (iIL-6) response in monocytes. Whole blood was incubated for 2 h at 37.0 °C, 38.5 °C and 40.0 °C, in the absence or presence of 0.5 µg/ml eHsp72. Flow cytometry was used to assess iHsp72 and iIL-6 expression in total monocytes and the three monocyte subsets. Incubation at 40.0 °C (p < 0.001) but not 38.5 °C (p = 0.085) increased iHsp72 expression when compared with 37.0 °C, while there was no effect of temperature on iIL-6 expression (p = 0.635). Following incubation with eHsp72, the expression of iHsp72 in classical monocytes was reduced at all temperatures (p < 0.001), while there was no effect of eHsp72 on iIL-6 expression (p = 0.071). Large temperature elevations are needed to induce an acute iHsp72 response in monocytes. In addition, contrary to its suggested role as a danger signal for the innate immune system, eHsp72 reduced iHsp72 and iIL-6 expression in monocytes.


Subject(s)
Acute-Phase Reaction/immunology , HSP72 Heat-Shock Proteins/metabolism , Hot Temperature , Interleukin-6/metabolism , Monocytes/immunology , Adult , HSP72 Heat-Shock Proteins/pharmacology , Healthy Volunteers , Humans , Interleukin-6/immunology , Male
3.
Vaccine ; 36(20): 2886-2895, 2018 05 11.
Article in English | MEDLINE | ID: mdl-29627234

ABSTRACT

In this study, the potential of N-trimethyl chitosan (TMC) nanoparticles as a carrier system for the nasal delivery of the r4M2e.HSP70c, as an M2e-based universal recombinant influenza virus vaccine candidate, was investigated in mice. The anti-M2e specific cellular and humoral immune responses were assessed and the protective efficacy against a 90% lethal dose (LD90) of influenza A/PR/8/34 (H1N1) in a mice model was evaluated. Our results showed that the intranasal immunization of mice with r4M2e.HSP70c+TMC rather than the control groups, r4M2e+TMC, r4M2e and PBS (Phosphate buffer saline), significantly elevated both longevity and serum level of the total M2e-specific IgG antibody with a significant shift in the IgG2a/IgG1 ratio toward IgG2a, induced a Th1 skewed humoral and cellular immune responses, increased IFN-γ, IgG, and IgA in the bronchoalveolar lavage fluid (BALF), and promoted the proliferation of peripheral blood lymphocytes with lower morbidity and mortality rate against viral challenge. In conclusion, based on evidence to our finding, nasal vaccination with r4M2e.HSP70c antigen encapsulated into N-Trimethyl Chitosan (TMC) nanoparticulate system showed to induce a long lasting M2e-specific humoral and cellular immune responses and also provided full protection against a 90% lethal dose (LD90) of the influenza virus A/PR/8/34 (H1N1). It seems, protective immunity following intranasal administration of r4M2e could be resulted by the cooperation of both adjuvants, TMC and HSP70c.


Subject(s)
Adjuvants, Immunologic/pharmacology , Chitosan/administration & dosage , Drug Carriers/administration & dosage , HSP72 Heat-Shock Proteins/pharmacology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Viral Matrix Proteins/immunology , Adjuvants, Immunologic/administration & dosage , Administration, Intranasal , Animals , Antibodies, Viral/analysis , Bronchoalveolar Lavage Fluid/immunology , Cell Proliferation , Disease Models, Animal , Female , HSP72 Heat-Shock Proteins/administration & dosage , Humans , Immunity, Cellular , Immunity, Humoral , Immunoglobulin A/analysis , Immunoglobulin G/analysis , Influenza, Human/prevention & control , Interferon-gamma/analysis , Leukocytes, Mononuclear/immunology , Mice, Inbred BALB C , Nanoparticles/administration & dosage , Serum/immunology , Survival Analysis , Treatment Outcome , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology , Viral Matrix Proteins/administration & dosage
4.
Arch Oral Biol ; 83: 181-186, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28783551

ABSTRACT

OBJECTIVE: The aim of the present study was to examine the effect of extracellular heat shock protein (HSP) 72 on human periodontal ligament fibroblast cells (hPDLFs) in vitro. DESIGN: hPDLFs were stimulated by recombinant human HSP72 (rhHSP72). TAK-242 was used to inhibit toll-like receptor 4 (TLR4) activity. Interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF)-α mRNA levels were analyzed by real-time PCR and protein levels were analyzed by enzyme-linked immunosorbent assay. p65/RelA phosphorylation was analyzed by western blot. RESULTS: IL-6, IL-8 and TNF-α mRNA and protein levels were significantly increased by rhHSP72 stimulation. These effects were inhibited by TAK-242 treatment. Additionally, p65/RelA phosphorylation was increased after 5-min rhHSP72 stimulation, which was inhibited by TAK-242 treatment. CONCLUSION: Extracellular HSP72 induces proinflammatory cytokines through TLR4/NF-κB in hPDLFs.


Subject(s)
Fibroblasts/metabolism , HSP72 Heat-Shock Proteins/pharmacology , Interleukin-6/metabolism , Interleukin-8/metabolism , NF-kappa B/antagonists & inhibitors , Periodontal Ligament/cytology , Toll-Like Receptor 4/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Phosphorylation , Real-Time Polymerase Chain Reaction
5.
Microb Pathog ; 111: 132-138, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28843722

ABSTRACT

Targeted immune therapies are a modern approach to harness the immunity to treat cancer patients. Exosomes (EXOs) are nano-vesicles used for drug delivery in cancer treatment. We aimed to assess the effectiveness of novel designed EXO structures for immunotherapy alone and in combination with other components in animal models. EXO derived from untreated macrophage (EXO), WEHI-164 cell lysate treated EXO (EXOLys), HSP70 enriched WEHI-164 cell lysate treated EXO (EXOHSP70), Naloxone (NLX) treated EXO (EXONLX), Propranolol (PRP) treated EXO (EXOPRP) and staphylococcal enterotoxin B (SEB) anchored to three kinds of EXOs designated as EXO/SEB, EXOLys/SEB, EXOHSP70/SEB were purified from J774 cell line. To determine the therapeutic effect of these novel constructed nano-vesicles, the animals were immunized with different types of EXOs at weekly intervals for three consecutive weeks and in the fourth week the WEHI-164 tumor cells were injected. Finally, the splenocyte proliferation was examined by MTT assay and tumor growth was also determined in each group. We observed that EXOHSP was more effective than EXO and EXOLys to decrease the number of tumor cells and to stimulate immune responses in animal models (P < 0.05). In SEB-anchored EXO group, EXOHSP70/SEB has the potency to stimulate immune responses more efficiently than EXO/SEB and EXOLys/SEB and the tumor was not palpable until 28th day which may refer to synergistic effect of HSP70 and SEB on immunity. In EXONLX treated mice proliferative response decreased significantly compared to control group (P > 0.05) and the tumor number was constant within a period of 28 days and EXOPRP may delay the occurrence of the fibrosarcoma tumor; After development of fibrosarcoma the number of tumors diminished over the studied period of time. Our results demonstrate that HSP70 enriched EXO is an effective immunoadjuvant in cancer immunotherapy and causes tumor regression in animal model.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems/methods , Enterotoxins/pharmacology , Exosomes , Fibrosarcoma/prevention & control , Immunotherapy/methods , Macrophages , Adaptive Immunity , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Fibrosarcoma/pathology , HSP72 Heat-Shock Proteins/pharmacology , Immunization , Iran , Male , Mice , Mice, Inbred BALB C , Models, Animal , Naloxone/pharmacology , Vaccination/methods
6.
Biomed Res Int ; 2015: 508101, 2015.
Article in English | MEDLINE | ID: mdl-26221596

ABSTRACT

This study examined the role of exogenous heat shock protein 72 (Hsp72) in reversing sepsis-induced liver dysfunction. Sepsis was induced by cecal ligation and puncture. Liver function was determined on the basis of the enzymatic activities of serum glutamate oxaloacetate transaminase (GOT) and glutamate pyruvate transaminase (GPT). Apoptosis was determined using terminal deoxynucleotidyl transferase dUTP nick end labeling staining. B-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (Bax), cleaved caspase-3 and caspase-9, and cleaved poly (ADP-ribose) polymerase (PARP) protein expressions were analyzed using Western blotting. Results showed GOT and GPT levels increased during sepsis, and levels were restored following the administration of human recombinant Hsp72 (rhHsp72). Increased liver tissue apoptosis was observed during sepsis, and normal apoptosis resumed on rhHsp72 administration. The Bcl-2/Bax ratio, cleaved caspase-3, caspase-9, and PARP protein expressions in the liver tissues were upregulated during sepsis and normalized after rhHsp72 treatment. We conclude that, during sepsis, exogenous Hsp72 restored liver dysfunction by inhibiting apoptosis via the mitochondria-initiated caspase pathway.


Subject(s)
HSP72 Heat-Shock Proteins/therapeutic use , Liver Diseases/complications , Liver Diseases/drug therapy , Sepsis/complications , Sepsis/drug therapy , Animals , Apoptosis/drug effects , HSP72 Heat-Shock Proteins/pharmacology , Humans , Liver Diseases/pathology , Male , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Sepsis/pathology , bcl-2-Associated X Protein/metabolism
8.
J Physiol Biochem ; 69(1): 125-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22798210

ABSTRACT

Regular physical exercise is recognized as a nonpharmacological therapeutic strategy in the treatment of metabolic syndrome, and has been proposed for improving obesity, diabetic status, insulin resistance, and immune response. The aim of the present study was to evaluate the effect of a regular exercise program (treadmill running, 5 days/week for 14 weeks at 35 cm/s for 35 min in the last month) on the release of the pro-inflammatory cytokine interferon gamma (IFNγ) by peritoneal cells (macrophages and lymphocytes) from obese Zucker rats (fa/fa) in response to noradrenaline (NA) and heat shock proteins of 72 kDa (Hsp72), and the possible adaptation due to training for a bout acute exercise (a single session of 25-35 min at 35 cm/s). In healthy (lean Fa/fa) and obese animals, peritoneal cells released greater concentrations of IFNγ in response to Hsp72 and lower concentrations in response to NA. The regular exercise training protocol, evaluated in the obese animals, produced a clear change in the regulation of the release of IFNγ. Peritoneal immune cells from trained animals released more IFNγ in response to NA, but there was a reduction in the release of IFNγ in response to Hsp72. In the obese animals, regular exercise caused a change in the inhibitory effect of NA (which now becomes stimulatory) and the stimulatory effect of Hsp72e (which now becomes inhibitory) in relation to the release of IFNγ. This reflects that Hsp72, induced by the prior release of NA following exercise-induced stress, plays a role in the homeostatic balance of release of IFNγ by peritoneal immune cells in obese animals during exercise.


Subject(s)
HSP72 Heat-Shock Proteins/pharmacology , Interferon-gamma/metabolism , Lymphocytes/drug effects , Macrophages, Peritoneal/drug effects , Metabolic Syndrome/pathology , Norepinephrine/pharmacology , Obesity/pathology , Animals , Interferon-gamma/biosynthesis , Lymphocytes/immunology , Lymphocytes/pathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/pathology , Male , Metabolic Syndrome/immunology , Obesity/immunology , Physical Conditioning, Animal , Primary Cell Culture , Rats , Rats, Zucker
9.
Article in Chinese | MEDLINE | ID: mdl-23156730

ABSTRACT

OBJECTIVE: To investigate the effects of heat shock protein 72 (Hsp72) on the expression of IL-6 and IL-8 and activation of NF-kappaB in synoviocytes from patients suffered from rheumatoid arthritis (RA). METHODS: IL6 and IL8 concentrations in culture supernatants were measured using enzyme-linked immunosorbent assays (ELISA). Nuclear translocation of NF-kappaB and degradation of the inhibitory protein IkappaBalpha were examined using immunohistochemistry and Western blot. RESULTS: Hsp72 down-regulated IL-6 and IL-8 production in RA synoviocytes induced by tumor necrosis factor-alpha (TNF-alpha). Hsp72 inhibited nuclear translocation of NF-kappaB and degradation of IkappaBalpha induced by TNF-alpha. CONCLUSION: Hsp72 has an anti-inflammatory effect on RA by down-regulation of IL-6 and IL-8 in synoviocytes, which is mediated through inhibiting the activation of NF-KalphaB signal pathways.


Subject(s)
Arthritis, Rheumatoid/metabolism , HSP72 Heat-Shock Proteins/pharmacology , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Cells, Cultured , Humans , I-kappa B Proteins/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Signal Transduction , Synovial Membrane/cytology , Tumor Necrosis Factor-alpha/pharmacology
10.
J Neurosci Res ; 90(6): 1261-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22488728

ABSTRACT

The cytoprotective role of extracellular heat shock protein (Hsp) 72 has been demonstrated in various cell types, including neuronal cells; however, few studies have investigated the actual role of Hsp72 in the survival of Schwann cells (SCs). In the present study, we investigated the effect of exogenous Hsp72 on Schwann cell apoptotic cell death induced by H2O2. We determined that extracellular exposure to Hsp72 reduced cell death in rat SCs in a dose-dependent manner, with the protection resulting from downregulation of apoptosis induced by H2O2 (as shown by TUNEL and annexin V flow cytometry analyses). Moreover, we observed that Hsp72 suppressed caspase-3 and -9 activation induced by H2O2. This was accompanied by upregulation of the antiapoptotic protein Bcl-2. These findings indicate that extracellular Hsp72 can afford neuroprotection to peripheral nerves via its ability to inhibit Schwann cell apoptosis and diminish oxidative stress-mediated injuries.


Subject(s)
Apoptosis/drug effects , HSP72 Heat-Shock Proteins/pharmacology , Hydrogen Peroxide/pharmacology , Oxidants/pharmacology , Schwann Cells/drug effects , Animals , Annexin A5/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Transformed , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Interactions , In Situ Nick-End Labeling/methods , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Tetrazolium Salts , Thiazoles , Up-Regulation/drug effects
11.
Am J Pathol ; 179(6): 2822-34, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22001349

ABSTRACT

Peritoneal dialysis-related peritonitis causes the denudation of mesothelial cells and, ultimately, membrane integrity alterations and peritoneal dysfunction. Because heat shock protein 72 (HSP72) confers protection against apoptosis and because autophagy mediates survival in response to cellular stresses, we examined whether autophagy contributes to HSP72-mediated cytoprotection in lipopolysaccharide (LPS)-induced peritonitis. Exposure of cultured peritoneal mesothelial cells to LPS resulted first in autophagy and later, apoptosis. Inhibition of autophagy by 3-methyladenine or Beclin-1 small-interfering RNA sensitized cells to apoptosis and abolished the antiapoptotic effect of HSP72, suggesting that autophagy activation acts as a prosurvival mechanism. Overexpression of HSP72 augmented autophagy through c-Jun N-terminal kinase (JNK) phosphorylation and Beclin-1 up-regulation. Suppression of JNK activity reversed HSP72-mediated Beclin-1 up-regulation and autophagy, indicating that HSP72-mediated autophagy is JNK dependent. In a rat model of LPS-associated peritonitis, autophagy occurred before apoptosis in peritoneum. Up-regulation of HSP72 by geranylgeranylacetone increased autophagy, inhibited apoptosis, and attenuated peritoneal injury, and these effects were blunted by down-regulation of HSP72 with quercetin. Additionally, blocking autophagy by chloroquine promoted apoptosis and aggravated LPS-associated peritoneal dysfunction. Thus, HSP72 protects peritoneum from LPS-induced mesothelial cells injury, at least in part by enhancing JNK activation-dependent autophagy and inhibiting apoptosis. These findings imply that HSP72 induction might be a potential therapy for peritonitis.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , HSP72 Heat-Shock Proteins/pharmacology , Lipopolysaccharides/toxicity , Peritonitis/chemically induced , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Apoptosis Regulatory Proteins/pharmacology , Beclin-1 , Cell Survival/drug effects , Cells, Cultured , Chloroquine/pharmacology , Diterpenes/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Green Fluorescent Proteins/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Luminescent Agents/metabolism , MAP Kinase Signaling System/drug effects , Male , Peritoneal Cavity/cytology , Peritonitis/prevention & control , Phosphorylation , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Up-Regulation
12.
Immunol Lett ; 135(1-2): 173-9, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21094186

ABSTRACT

Heat shock protein 72 (Hsp72), a canonical intracellular molecular chaperone, may also function as an extracellular danger signal for the innate immune system. To further delineate the biological role of Hsp72 in the innate immune system, we generated two truncated versions of the full length human Hsp72 (N-terminus Hsp72, amino acids 1-430; and C-terminus Hsp72 amino acids 420-641) and directly compared their ability to activate cells from the macrophage/monocyte lineage. In RAW 264.7 macrophages transfected with a NF-κB-dependent luciferase reporter plasmid, C-terminus Hsp72 was a more potent inducer of NF-κB activity than N-terminus Hsp72, and this effect did not seem to be secondary to endotoxin contamination. C-terminus Hsp72-mediated activation of the NF-κB pathway was corroborated by increased activation of IκB kinase, degradation of IκBα, and increased NF-κB-DNA binding. C-terminus Hsp72 was a more potent inducer of tumor necrosis factor-α (TNFα) expression in RAW 264.7 macrophages and in primary murine peritoneal macrophages from wild-type mice. C-terminus Hsp72 did not induce TNFα expression in primary murine peritoneal macrophages from Toll-like receptor (TLR4) mutant mice, indicating a role for TLR4. In human THP-1 mononuclear cells, C-terminus Hsp72 induced tolerance to subsequent LPS stimulation, whereas N-terminus Hsp72 did not induce tolerance. Finally, control experiments using equimolar amounts of N-terminus or C-terminus Hsp72 demonstrated a higher biological potency for C-terminus Hsp72. These data demonstrate that the ability of human Hsp72 to serve as an activator for cells of the macrophage/monocyte lineage primarily lies in the C-terminus region spanning amino acids 420-641.


Subject(s)
HSP72 Heat-Shock Proteins/immunology , Immunity, Innate/immunology , Macrophage Activation/immunology , Macrophages, Peritoneal/immunology , Monocytes/immunology , Animals , Cell Line , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/pharmacology , Humans , Immunity, Innate/drug effects , Macrophage Activation/drug effects , Macrophages, Peritoneal/metabolism , Mice , Mice, Mutant Strains , Monocytes/metabolism , Protein Structure, Tertiary , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 4/metabolism
13.
J Huazhong Univ Sci Technolog Med Sci ; 30(3): 337-43, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20556578

ABSTRACT

The study investigated the effects of heat shock protein 70 (HSP70) antisense oligonucleotide (ASODN) on the proliferation and apoptosis of a human hepatocellular carcinoma cell line (SMMC-7721 cells) in vitro. HSP70 oligonucleotide was transfected into SMMC-7721 cells by the mediation of Sofast transfection reagent. Inhibition rate of SMMC-7721 cells was determined by using MTT method. Apoptosis rate and cell cycle distribution were measured by flow cytometry. Immunocytochemistry staining was used to observe the expression of HSP70, Bcl-2 and Bax. The results showed that HSP70 ASODN at various concentrations could significantly inhibit the growth of SMMC-7721 cells, and the inhibition effect peaked 48 h after transfection with 400-nmol/L HSP70 ASODN. Cytometric analysis showed the apoptotic rate was increased in a dose- and time-dependent manner in the HSP70 ASODN-treated cells. The percentage of cells in the G2/M and S phases was significantly decreased and that in the G0/G1 phase increased as the HSP70 ASODN concentration was elevated and the exposure time prolonged. Immunocytochemistry showed that treatment of SMMC-7721 cells with HSP70 ASODN resulted in decreased expressions of HSP70 and Bcl-2 proteins, and an increased expression of Bax protein. It was concluded that the HSP70 ASODN can inhibit the growth of the SMMC-7721 cells and increase cell apoptosis by down-regulating the expression of HSP70. HSP70 ASODN holds promise for the treatment of hepatocellular carcinoma.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , HSP72 Heat-Shock Proteins/genetics , Liver Neoplasms/pathology , Oligonucleotides, Antisense/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , HSP72 Heat-Shock Proteins/pharmacology , Humans
14.
J Biol Chem ; 285(1): 349-56, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19861412

ABSTRACT

Extracellular heat shock protein 72 (Hsp72; inducible form of the 70-kDa heat shock protein) plays a critical role in innate and adaptive immune responses and has shown promise as an ideal adjuvant for the optimization of antigen-specific anti-tumor vaccines. Recent studies suggest that to correctly elucidate the mechanisms by which Hsp72 exerts its beneficial effects in vitro, great care must be taken to ensure that endotoxin by-products do not invalidate the findings. In this study, we have taken advantage of the baculovirus expression vector system for production of endotoxin-free recombinant Hsp72. The coding sequence of human hsp72 was recombined into the baculovirus immediately downstream of the strong polyhedron gene promoter. Ninety-six h post-infection of Sf9 insect cells with recombinant baculovirus, maximal levels of Hsp72 protein were detected. The recombinant human Hsp72 was purified by affinity chromatography from insect cells, and purity was confirmed by SDS-PAGE and mass spectrometry. The purified human recombinant Hsp72(bv) (Hsp72 produced using the BEVS) was demonstrated to have no endotoxin contamination and was shown to have stimulated potent calcium flux in the human monocytic cell line. Furthermore, recombinant Hsp72(bv) enhanced the tolerance of neuroblastoma cells to heat stress-induced cell death and displayed classical chaperokine functions including augmentation of inflammatory cytokine productions in mouse splenocytes. The production of functional, endotoxin-free recombinant human Hsp72(bv) in insect cells is inexpensive and convenient and eliminates the need of special procedures for endotoxin depletion. Endotoxin-free recombinant human Hsp72(bv) can now be used to unlock the important role Hsp72 plays in modulating immune function.


Subject(s)
Baculoviridae/genetics , HSP72 Heat-Shock Proteins/metabolism , Insecta/cytology , Recombinant Proteins/metabolism , Animals , Baculoviridae/drug effects , Calcium/metabolism , Cell Death/drug effects , Cell Line , Cytokines/biosynthesis , Cytoprotection/drug effects , Genetic Vectors/genetics , HSP72 Heat-Shock Proteins/genetics , HSP72 Heat-Shock Proteins/isolation & purification , HSP72 Heat-Shock Proteins/pharmacology , Heat-Shock Response/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Leukocytes/cytology , Leukocytes/drug effects , Mice , Neuroblastoma/pathology , Phenotype , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/metabolism
15.
Vaccine ; 27(6): 870-81, 2009 Feb 05.
Article in English | MEDLINE | ID: mdl-19084567

ABSTRACT

APOBEC3G is an innate intracellular anti-viral factor which deaminates retroviral cytidine to uridine. In vivo studies of APOBEC3G (A3G) were carried out in rhesus macaques, following mucosal immunization with SIV antigens and CCR5 peptides, linked to the 70kDa heat shock protein. A progressive increase in A3G mRNA was elicited in PBMC after each immunization (p<0.0002 to p< or =0.02), which was maintained for at least 17 weeks. Analysis of memory T cells showed a significant increase in A3G mRNA and protein in CD4(+)CCR5(+) memory T cells in circulating (p=0.0001), splenic (p=0.0001), iliac lymph nodes (p=0.002) and rectal (p=0.01) cells of the immunized compared with unimmunized macaques. Mucosal challenge with SIVmac 251 showed a significant increase in A3G mRNA in the CD4(+)CCR5(+) circulating cells (p<0.01) and the draining iliac lymph node cells (p<0.05) in the immunized uninfected macaques, consistent with a protective effect exerted by A3G. The results suggest that mucosal immunization in a non-human primate can induce features of a memory response to an innate anti-viral factor in CCR5(+)CD4(+) memory and CD4(+)CD95(+)CCR7(-) effector memory T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytidine Deaminase/biosynthesis , Mucous Membrane , SAIDS Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Cells, Cultured , HSP72 Heat-Shock Proteins/administration & dosage , HSP72 Heat-Shock Proteins/pharmacology , Lymph Nodes/immunology , Macaca mulatta , Receptors, CCR5/administration & dosage , Rectum/immunology , SAIDS Vaccines/administration & dosage , Spleen/immunology
16.
Am J Physiol Cell Physiol ; 295(2): C514-20, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18508912

ABSTRACT

Heat shock protein (HSP) 72 is released by cells during stress and injury. HSP-72 also stimulates the release of cytokines in macrophages by binding to Toll-like receptors (TLR) 2 and 4. Circulating levels of HSP-72 increase during hepatic ischemia-reperfusion injury. The role of extracellular HSP-72 (eHSP-72) in the injury response to ischemia-reperfusion is unknown. Therefore, the objective of the present study was to determine whether eHSP-72 has any direct effects on hepatocytes. Primary mouse hepatocytes were treated with purified human recombinant HSP-72. Conditioned media were evaluated by ELISA for the cytokines, TNF-alpha, IL-6, and macrophage inflammatory protein 2 (MIP-2). Stimulation of hepatocytes with eHSP-72 did not induce production of TNFalpha or IL-6 but resulted in dose-dependent increases in MIP-2 production. To evaluate the pathway responsible for this response, expression of TLR2 and TLR4 was confirmed on hepatocytes by immunohistochemistry. Hepatocyte production of MIP-2 was significantly decreased in hepatocytes obtained from TLR2 or TLR4 knockout mice. MIP-2 production was found to be partially dependent on NF-kappaB because inhibition of NF-kappaB with Bay 11-7085 significantly decreased eHSP-72-induced MIP-2 production. Inhibitors of p38 mitogen-activated protein kinase or c-Jun NH(2)-terminal kinase had no effect on production of MIP-2 induced by eHSP-72. The data suggest that eHSP-72 binds to TLR2 and TLR4 on hepatocytes and signals through NF-kappaB to increase MIP-2 production. The fact that eHSP-72 did not increase TNF-alpha or IL-6 production may be indicative of a highly regulated signaling pathway downstream from TLR.


Subject(s)
HSP72 Heat-Shock Proteins/pharmacology , Hepatocytes/drug effects , Recombinant Proteins/pharmacology , Animals , Cells, Cultured , Chemokine CXCL2/metabolism , Enzyme Inhibitors/pharmacology , HSP72 Heat-Shock Proteins/genetics , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Immunohistochemistry , Interleukin-2/metabolism , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Liver/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Mutation , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitriles/pharmacology , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Sulfones/pharmacology , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
J Immunol ; 179(9): 6318-24, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17947709

ABSTRACT

Heat shock proteins are generally regarded as intracellular proteins acting as molecular chaperones; however, Hsp72 is also detected in the extracellular compartment. Hsp72 has been identified in the bronchoalveolar lavage fluid (BALF) of patients with acute lung injury. To address whether Hsp72 directly activated airway epithelium, human bronchial epithelial cells (16HBE14o-) were treated with recombinant Hsp72. Hsp72 induced a dose-dependent increase in IL-8 expression, which was inhibited by the NF-kappaB inhibitor parthenolide. Hsp72 induced activation of NF-kappaB, as evidenced by NF-kappaB trans-activation and by p65 RelA and p50 NF-kappaB1 binding to DNA. Endotoxin contamination of the Hsp72 preparation was not responsible for these effects. Next, BALB/c mice were challenged with a single intratracheal inhalation of Hsp72 and killed 4 h later. Hsp72 induced significant up-regulation of KC, TNF-alpha, neutrophil recruitment, and myeloperoxidase in the BALF. A similar challenge with Hsp72 in TLR4 mutant mice did not stimulate the inflammatory response, stressing the importance of TLR4 in Hsp72-mediated lung inflammation. Last, cultured mouse tracheal epithelial cells (MTEC) from BALB/c and TLR4 mutant and wild-type mice were treated ex vivo with Hsp72. Hsp72 induced a significant increase in KC expression from BALB/c and wild-type MTEC in an NF-kappaB-dependent manner; however, TLR4 mutant MTEC had minimal cytokine release. Taken together, these data suggest that Hsp72 is released and biologically active in the BALF and can regulate airway epithelial cell cytokine expression in a TLR4 and NF-kappaB-dependent mechanism.


Subject(s)
Cytokines/biosynthesis , HSP72 Heat-Shock Proteins/pharmacology , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Administration, Inhalation , Animals , Bronchi/metabolism , Cell Line , Epithelial Cells/metabolism , Epithelium/metabolism , Female , HSP70 Heat-Shock Proteins/metabolism , HSP72 Heat-Shock Proteins/administration & dosage , Humans , Inflammation/chemically induced , Inflammation/metabolism , Mice , Mice, Knockout , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
18.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 31(2): 228-31, 2006 Apr.
Article in Chinese | MEDLINE | ID: mdl-16706121

ABSTRACT

OBJECTIVE: To explore the protective effect of HSP72 on the acute injury of cardiomyocyte induced by oxidative stress. METHODS: Cardiomyocytes of neonatal rats treated with heat shock (42 degrees C, 30 min, recovery for 6 h) to induce the expression of HSP72 and HSP72 antisense oligonucleotide was transformed to block the expression of HSP72. 0.5 mmol/L (final concentration) H2O2 was added into the culture medium to mimic oxidative stress, and to induce the acute injury of neonatal cardiomyocytes. The release of LDH and the total protein synthesis were applied to evaluate the injury of cardiomyocyte of neonatal rats. RESULTS: Oxidative stress could significantly increase the release of LDH, and inhibit the total protein synthesis. By inducing the expression of HSPs, heat shock pretreatment significantly reduced the release of LDH and relieved the oxidative stress-mediated inhibition of total protein synthesis. Moreover, HSP7-2 anti-sense oligonucleotide could remarkably block the protective effect of heat shock pretreatment on the cellular injuries induced by H2O2. CONCLUSION: HSP72 plays a most important role in the acute injury of cardiomyocyte mediated by oxidative stress.


Subject(s)
HSP72 Heat-Shock Proteins/pharmacology , Myocytes, Cardiac/pathology , Oxidative Stress , Animals , Animals, Newborn , Cells, Cultured , L-Lactate Dehydrogenase/metabolism , Oligonucleotides, Antisense/pharmacology , Protein Biosynthesis , Random Allocation , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...